• 검색 결과가 없습니다.

Single intranasal immunization with recombinant adenovirus-based vaccine induces protective immunity against respiratory syncytial virus infection

N/A
N/A
Protected

Academic year: 2021

Share "Single intranasal immunization with recombinant adenovirus-based vaccine induces protective immunity against respiratory syncytial virus infection"

Copied!
8
0
0

로드 중.... (전체 텍스트 보기)

전체 글

(1)

0022-538X/08/$08.00

⫹0 doi:10.1128/JVI.02372-07

Copyright © 2008, American Society for Microbiology. All Rights Reserved.

Single Intranasal Immunization with Recombinant Adenovirus-Based

Vaccine Induces Protective Immunity against Respiratory

Syncytial Virus Infection

Jae-Rang Yu, Sol Kim, Jee-Boong Lee, and Jun Chang*

Division of Life and Pharmaceutical Sciences, Ewha Womans University, 11-1 Dae-Hyun Dong,

Seo-Dae-Mun Gu, Seoul 120-750, Korea

Received 2 November 2007/Accepted 6 December 2007

Respiratory syncytial virus (RSV) is a major cause of severe lower respiratory tract disease in infancy and

early childhood. Despite its importance as a pathogen, there is no licensed vaccine against RSV. The G

glycoprotein of RSV, a major attachment protein, is a potentially important target for protective antiviral

immune responses. Here, a recombinant replication-deficient adenovirus-based vaccine, rAd/3xG, expressing

the soluble core domain of G glycoprotein (amino acids 130 to 230) engineered by codon optimization and

tandem repetition for higher-level expression, was constructed and evaluated for its potential as an RSV

vaccine in a murine model. A single intranasal immunization with rAd/3xG provided potent protection against

RSV challenge which lasted for more than 10 weeks. Strong mucosal immunoglobulin A responses were also

induced by a single intranasal immunization but not by intramuscular or oral administration of rAd/3xG.

Interestingly, neither gamma interferon- nor interleukin-4-producing CD4 T cells directed to I-E

d

-restricted

epitope were detected in the lungs of rAd/3xG-immune mice upon challenge, whereas priming with vaccinia

virus expressing RSV G (vvG) elicited strong Th1/Th2 mixed CD4 T-cell responses. Lung eosinophilia and

vaccine-induced weight loss were significantly lower in the rAd/3xG-immune group than in the vvG-primed

group. Together, our data demonstrate that a single intranasal administration of rAd/3xG elicits beneficial

protective immunity and represents a promising vaccine regimen against RSV infection.

Respiratory syncytial virus (RSV) is the most important viral

pathogen causing serious respiratory tract disease in infants

and young children worldwide. RSV is also receiving

increas-ing recognition as an important cause of lower respiratory tract

illness in immunocompromised patients and the elderly (13,

15, 16). Despite the importance of RSV as a respiratory

patho-gen, there is no licensed vaccine currently available against

RSV infection. Thus, developing an effective and safe RSV

vaccine remains a worldwide priority.

The RSV G glycoprotein was identified as the major RSV

attachment protein (24) and is thought to be important for

protection against RSV infection (39). G protein lacks any

major histocompatibility complex class I-restricted epitope (8,

26, 36) and has not yet been demonstrated to elicit a cytotoxic

T-lymphocyte response in either humans or mice (19, 29). It

has a single immunodominant I-E

d

epitope spanning amino

acids 183 to 198 and largely induces a specific subset of CD4 T

cells restricted to V

␤14 expression in the T-cell receptor (40,

42). Numerous studies have suggested that immunization with

RSV G is associated with the induction of polarized Th2-type

responses, which leads to pulmonary eosinophilia upon RSV

challenge of G-immunized mice (17, 20, 30, 35, 40). In

con-trast, it was recently suggested that G-specific immune

re-sponses are not solely the basis for vaccine-enhanced illness

and should not be excluded from potential vaccine strategies

(21, 22). In addition, intramuscular (i.m.) injection of plasmid

DNA encoding membrane G or secreted G induced balanced

Th1/Th2 immunity without an atypical pulmonary

inflamma-tion after RSV challenge in mice and cotton rats (3, 25),

suggesting that G protein may provide protection but not

in-duce enhanced lung pathology, depending on the vehicle

and/or method of delivery.

In the present study, we have targeted the RSV G protein

fragment between residues 130 and 230 and engineered the

sequence by codon optimization for optimal expression in

an-imal cells and by the addition of a secretory signal sequence

derived from tissue plasminogen activator (t-PA). Moreover,

this sequence was engineered to be multiple-copy tandem

re-peats in the same open reading frame for higher

immunoge-nicity (23, 31, 47). Replication-defective recombinant

adeno-virus (rAd) vaccines (rAd/1xG and rAd/3xG) were then

generated and evaluated for their potential as vaccines. We

show here that a single intranasal (i.n.) immunization of rAd/

3xG induces a strong serum immunoglobulin G (IgG)

re-sponse, a mucosal IgA rere-sponse, and long-term protection

following RSV challenge without vaccine-enhanced illness.

MATERIALS AND METHODS

Preparation of RSV stock.RSV strain A2 was propagated in HEp-2 cells (ATCC, Manassas, VA) in Dulbecco’s modified Eagle’s medium (Life Technol-ogies, Gaithersburg, MD) supplemented with 3% heat-inactivated fetal calf serum, 2 mM glutamine, 20 mM HEPES, nonessential amino acid, penicillin, and streptomycin and titrated for infectivity by plaque assay.

Construction of replication-defective rAds.A coding sequence of RSV G protein spanning amino acid residues 130 to 230 (RSV strain A2) was synthe-sized, in which codon substitutions were made for minimized usage of rare codons (Bioneer Corp., Daejeon, Korea). Codons for a six-histidine stretch as a

* Corresponding author. Mailing address: College of Pharmacy,

Ewha Womans University, 11-1 Dae-Hyun Dong, Seo-Dae-Mun Gu,

Seoul 120-750, Korea. Phone: 82-2-3277-2549. Fax: 82-2-3277-3051.

E-mail: tcell@ewha.ac.kr.

Published ahead of print on 19 December 2007.

2350

on March 16, 2017 by Ewha Woman`s University

http://jvi.asm.org/

(2)

tag and two consecutive stop codons were attached at 3⬘ terminus. This synthetic DNA was sequenced and then subcloned into pGEM-t-EASY vector (Promega, Madison, WI). The RSV G protein region was again amplified with various oligonucleotide primers containing appropriate restriction enzyme sites and jux-taposed three times with four glycine residues as a linker between each repeated region (Fig. 1). The original RSV G sequences were also cloned into the same sites of the vector. A start codon and the signal sequence of human t-PA were then inserted in the N termini of the RSV G fragments. Finally, the entire open reading frame was excised by KpnI/XhoI digestion and inserted into the same sites of pShuttle-CMV vector. Replication-defective Ads (serotype 5) were gen-erated by insertion of foreign sequences by homologous recombination and subsequent purification of recombinant progeny as described previously (18). Briefly, the shuttle vector plasmid was electroporated into electrocompetent BJ5183 cells carrying the pAdEasy-1 Ad genomic DNA. Recombinant Ad DNA was isolated and transfected into HEK293 cells to generate rAd/1xG and rAd/ 3xG viruses. Control Ad (rAd/control) was generated by the same method using the empty pShuttle-CMV vector. The recombinant viruses were amplified, pu-rified on CsCl2gradients, and titrated according to the protocols provided by the

manufacturer. The correct expression and secretion of RSV G protein fragments by rAd/3xG-infected HEp-2 cells were verified by Western blotting using G-specific monoclonal antibody (clone 131-2G; Chemicon International, Inc., Temecula, CA) and horseradish peroxidase-conjugated streptavidin (Zymed Laboratories, San Francisco, CA).

Immunization and challenge. Female BALB/c mice were purchased from Charles River Laboratories Inc. (Yokohama, Japan) and kept under specific-pathogen-free conditions. For immunization, 6- to 8-week-old mice were inoc-ulated with various doses of replication-defective Ad via the i.n., i.m., or oral route. For i.n. immunizations, mice were lightly anesthetized by ether/chloroform inhalation, and 5⫻ 107

PFU vaccine or control virus in a volume of 50␮l was applied to the left nostril. i.m. immunization was performed by injection of 5⫻ 107

PFU of vaccine in 100␮l into mouse hind limbs. For oral immunization, mice were deprived of water and food a few hours before injection and immunized with 5⫻ 107

PFU of virus in 200␮l of phosphate-buffered saline (PBS) by proximal esophageal intubation with a mouse feeding needle. Male and female pups were immunized with 3⫻ 107

PFU of rAd in volumes of 20 to 30␮l at 7 to 14 days of age. Vaccinia virus expressing RSV G (vvG) (5⫻ 106PFU) was

inoculated at the base of the tail by scarification (36). Three to four weeks later the mice were challenged i.n. with 3⫻ 106to 1⫻ 107PFU of live RSV A2 if

necessary. All animal studies were performed according to the guidelines of our Institutional Animal Care and Use Committee.

ELISA.Blood was obtained from the retro-orbital plexus with a heparinized capillary tube, collected in an Eppendorf tube, and centrifuged, and serum was stored at⫺20°C. RSV G protein-specific antibody titers in immunized mice were measured by a direct enzyme-linked immunosorbent assay (ELISA). Briefly, 96-well plates were coated overnight with 100␮l/well of 10-␮g/ml RSV A2 antigen (U.S. Biological, Swampscott, MA) or 2⫻ 104PFU of purified RSV A2

diluted in PBS and then blocked with PBS containing 2% bovine serum albumin and 0.05% Tween 20 for 2 h. Sera or lavage fluids were then added in serial dilutions and incubated for 2 h. The plates were washed five times with PBS containing 0.05% Tween 20 and incubated for 1 h with various dilutions of horseradish peroxidase-conjugated affinity-purified rabbit anti-mouse total IgG,

IgG1, IgG2a, or IgA secondary antibody (Zymed Laboratories, San Francisco, CA). The plates were washed three times and developed with 3,3⬘,5,5⬘-tetra-methylbenzidine, and the reaction was stopped with 1 M H3PO4and analyzed at

450 nm with a Thermo ELISA plate reader.

RSV titer in lung. Four days after RSV challenge, subsets of mice were euthanized and the lungs were removed into Eagle’s modified essential medium. The tissues were then processed through a steel screen to obtain a single-cell suspension, and particulate matter was removed by passage through a 70-␮m cell strainer (BD Labware, Franklin Lakes, NJ). The supernatants were collected, and RSV titers in the supernatants were measured by plaque assay on subcon-fluent HEp-2 monolayers. The data are expressed as the PFU per gram of lung tissue.

BAL.At 5 days postchallenge, a subset of mice from each group were sacrificed and tracheotomy was performed. The lung airways were washed with 0.8 ml of PBS containing 1% fetal bovine serum (FBS). The collected bronchoalveolar lavage (BAL) fluid cells and supernatants were used for counting eosinophils by hematoxylin-eosin staining and measuring secretory IgA titers, respectively.

Preparation of lung lymphocytes and flow cytometric analysis.The lungs were perfused with 5 ml of PBS containing 10 U/ml heparin (Sigma-Aldrich, St. Louis, MO) through the right ventricle using a syringe fitted with 25-gauge needle. The lungs were then removed and placed in RPMI medium supplemented with glutamine, gentamicin, penicillin G, and 10% FBS (HyClone, Logan, UT). The tissue was then processed through a steel screen to obtain a single-cell suspen-sion, and particulate matter was removed by passage through a 70-␮m Falcon cell strainer (BD Labware, Franklin Lakes, NJ). Freshly explanted BAL fluid or lung cells were purified by density gradient centrifugation and stained in PBS–3% FBS–0.09% NaN3using fluorochrome-conjugated antibodies. The antibodies

used were anti-CD3e (clone 145-2C11), anti-CD4 (clone RM4-5), anti-CD44 (clone IM7), anti-CD49d (clone R1-2), and anti-V␤14 TCR (clone 14-2). All antibodies were purchased from BD PharMingen (San Diego, CA). After stain-ing, cells were fixed in PBS–2% (wt/vol) paraformaldehyde, and events were acquired using a FACSCalibur flow cytometer (BD Biosciences, San Diego, CA). To enumerate the number of cytokine-producing cells, intracellular cytokine staining was performed. In brief, 2⫻ 106

freshly explanted lung lymphocytes were cultured in a culture tube. Cells were left untreated or stimulated with 10 ␮M G(183–195) peptide (WAICKRIPNKKPG) and then incubated for 5 h at 37°C in 5% CO2. Brefeldin A (5␮g/ml) (Sigma-Aldrich) was added for the

duration of the culture period to facilitate intracellular cytokine accumulation. Cells were then stained for surface markers, washed, fixed, permeabilized with fluorescence-activated cell sorter buffer containing 0.5% saponin (Sigma-Aldrich, Seoul, Korea), and stained for gamma interferon (IFN-␥). The antibod-ies used were anti-IFN-␥ (clone XMG1.2) or its control isotype antibody (rat IgG1). Dead cells were excluded on the basis of forward and side light scatter patterns. Data were collected using CELLQuest software (BD Biosciences) and analyzed with CELLQuest and WinMDI version 2.9 software (Scripps Research Institute, La Jolla, CA).

Data analysis.Comparison of differences was conducted by using an unpaired, two-tailed Student t test. The difference was considered statistically significant when the P value wasⱕ0.05.

FIG. 1. Construction and characterization of rAd/3xG vaccine. (A) A shuttle vector was designed as shown in the diagram and constructed by

several cloning steps as described in Materials and Methods. The vector was used to generate the replication-deficient rAd/3xG by homologous

recombination with the Ad genomic DNA. (B) The expression of the recombinant RSV G protein fragment by rAd/3xG was confirmed by Western

blotting. Culture supernatants were prepared from HEp-2 cells infected with rAd/1xG, rAd/3xG, or rAd/control and subjected to Western blotting

with an anti-RSV glycoprotein monoclonal antibody. RSV A2-infected culture supernatant was used as a positive control.

V

OL

. 82, 2008

PROTECTIVE IMMUNITY AGAINST RSV INDUCED BY rAd VACCINE

2351

on March 16, 2017 by Ewha Woman`s University

http://jvi.asm.org/

(3)

RESULTS

Construction of rAd expressing an RSV G protein fragment.

The strategy that we employed to enhance the vaccine

poten-tial the of RSV G protein fragment (amino acid residues 130 to

230) was to increase expression by codon optimization and

secretion of the recombinant protein by the addition of a signal

sequence from t-PA. In addition, three copies of this RSV G

fragment were tandemly repeated for higher immunogenicity

(23, 31, 47). Given the natural tropism of Ad, a

replication-defective rAd vector might be an ideal candidate for delivery of

an RSV vaccine. Thus, the engineered coding sequence was

placed into Ad DNA under the control of a cytomegalovirus

enhancer and promoter by homologous recombination,

result-ing in the generation of the replication-defective rAd rAd/3xG

(Fig. 1A). For comparison, rAd/1xG, containing a single copy

of the RSV G fragment, was used. Using a G-specific

mono-clonal antibody and immunoblotting, multiple bands of

ap-proximately 20 kDa to 30 kDa were detected only in the

cul-ture supernatant from HEp-2 cells infected with rAd/3xG and

not in supernatant from either rAd/control- or

rAd/1xG-in-fected HEp-2 cells (Fig. 1B). However, we have observed a

weak positive signal in an ELISA with an rAd/1xG-infected

sample, indicating a much lower level of G fragment

expres-sion by rAd/1xG (data not shown). These results indicate that

rAd/3xG successfully expresses the specific RSV G protein

fragment and that the level of expression was significantly

enhanced by use of our strategy.

Humoral immune response to rAd/3xG.

We next examined

whether rAd/3xG vaccine was capable of eliciting

antigen-spe-cific immune responses in vivo and, if so, what were the

char-acteristics of such immunity and which route was the best one.

BALB/c mice were inoculated once via the i.n., i.m., or oral

route with the same doses of rAd/1xG, rAd/3xG, or control Ad.

As a comparison, live RSV A2 was given once by i.n.

instilla-tion, and at 2 weeks after immunization total anti-RSV IgG

titers were determined by ELISA. Both i.n. and i.m.

immuni-zation with rAd/3xG induced strong total IgG responses in

serum even after a single injection, as demonstrated in Fig. 2A.

FIG. 2. Characterization of humoral immune responses induced by rAd/3xG. (A) BALB/c mice were immunized once with rAd (rAd/1xG,

rAd/3xG, or rAd/control) or with live RSV A2 via the indicated routes, and systemic anti-RSV IgG antibody titers were measured by serum ELISA

3 weeks after immunization. The results represent log

2

end point values from four or five individual mice. (B) Average IgA titers in the BAL fluid

were measured 3 weeks after rAd/3xG immunization. Error bars indicate the standard deviation. (C) Total IgA titers in the supernatants of lung

homogenates were determined for each group 4 days after RSV challenge. (D) Fourteen-day-old pups were immunized with 3

⫻ 10

7

PFU of

rAd/3xG or rAd/control in a volume of 20

␮l. Anti-RSV IgG antibody titers were determined 2 weeks later.

on March 16, 2017 by Ewha Woman`s University

http://jvi.asm.org/

(4)

Oral administration led to a relatively weaker RSV-specific

serum IgG response than i.n. or i.m. application. However, i.n.

immunization with the same dose of rAd/1xG failed to elicit

any detectable IgG response above the control value (Fig. 2A).

Notably, the levels of serum IgG responses in

rAd/3xG-immu-nized mice (i.n.) were comparable to those induced by live

RSV inoculation (i.n.) (Fig. 2A), indicating potent

immunoge-nicity of the rAd/3xG vaccine in vivo.

Secretory IgA is the first line of host defense against aerial

pathogens and appears to protect against infection of upper

respiratory tract (6). Thus, an effective RSV vaccine should

induce virus-specific secretory IgA preferably in the respiratory

mucosal area. To examine whether rAd/3xG vaccination elicits

an IgA response in the respiratory tract, BAL was performed

after immunization and levels of IgA were determined by

RSV-specific ELISA. As shown in Fig. 2B, the level of mucosal

IgA was much higher in the i.n. immunized group than in the

other groups when BAL fluid was tested at 21 days after

im-munization. Each group of mice immunized i.m. or orally with

rAd/3xG exhibited basal levels of IgA response. We also

checked the level of anti-RSV IgA in the lungs at 4 days after

RSV challenge, and rAd/3xG-immune and RSV-immune mice

showed significantly higher levels of IgA than control mice

(P

⬍ 0.01) (Fig. 2C). The difference between the

rAd/3xG-immune and RSV-rAd/3xG-immune mice was not statistically

signifi-cant, although the level was slightly higher in rAd/3xG group.

Next, we investigated whether immunization with rAd/3xG

also induces strong humoral responses in young pups or

neo-nates. The 14-day-old pups immunized with rAd/3xG showed

anti-RSV IgG responses (Fig. 2D), although the mean titer was

lower than that in the adult group (Fig. 2A). However, we

failed to detect an anti-RSV humoral response in 7- and

10-day-old pups or neonates (data not shown), possibly due to

immaturity of the immune system (1).

We next examined the effect of preexisting immunity against

the Ad vector. The mice were preexposed to rAd/control,

rested for 3 weeks, and then immunized with the rAd/3xG

vaccine. There was no significant difference in the anti-RSV

antibody response between the nonpreexposed group and the

group preexposed to 1

⫻ 10

7

PFU (P

⫽ 0.11), although the

mean titer in the group preexposed to 1

⫻ 10

8

PFU was

decreased (P

⬍ 0.05) (Fig. 3). More interestingly, the levels of

secreted anti-RSV IgA in BAL fluid were not significantly

changed among all groups of mice (Fig. 3), and all groups of

mice showed no detectable RSV replication in the lungs when

challenged with 2

⫻ 10

6

PFU of RSV at 4 weeks after

immu-nization (data not shown). Taken together, our results suggest

that a single immunization with rAd/3xG vaccine via the i.n.

route is more efficient than that by other administration routes

in inducing both serum IgG and respiratory IgA and is

suffi-ciently immunogenic even in the presence of preexisting

anti-Ad immunity.

T-cell response to rAd/3xG.

It has been well demonstrated

that sensitization of mice with vvG primes G-specific CD4

T-cell responses and results in pulmonary eosinophilia

follow-ing RSV challenge (5, 17, 20, 30, 36). Since the G protein

fragment expressed by the rAd/3xG vaccine contains an I-E

d

-restricted CD4 T-cell epitope (43), we examined whether rAd/

3xG immunization primes G-specific CD4 T cells. To this end,

rAd/3xG- or vvG-immune mice were challenged with RSV,

and 4 days after challenge lung lymphocytes were prepared,

stimulated with I-E

d

-restricted G(183–195) epitope peptide,

and stained for IFN-

␥ production. As shown in Fig. 4A, a

G-specific CD4 T-cell response was barely detected in the

lungs of the rAd/3xG-immunized group (

ⱕ0.2% of lung CD4 T

cells), while a strong response (

ⱖ10% of lung CD4 T cells) was

observed in the vvG-immunized group by intracellular IFN-

staining. We also checked the level of interleukin-4 (IL-4)

cytokine after in vitro stimulation of lung mononuclear cells

from both groups. As shown in Fig. 4B, in vitro-stimulated

cells from vvG-immune mice produced much more IL-4 than

those from rAd/3xG-immune mice. Taken together, these

re-sults clearly demonstrate that rAd/3xG immunization does not

prime a significant G-specific Th1/Th2 response, in contrast to

vvG scarification.

It was previously reported that the CD4 T-cell response

directed to the I-E

d

-restricted immunodominant epitope

span-ning amino acids 183 to 195 of RSV G is sufficient to induce

enhanced disease after RSV challenge (40). Our results have

indicated that rAd/3xG immunization barely primes G-specific

CD4 T cells (Fig. 4A and B). Thus, it is quite unlikely that

rAd/3xG immunization would elicit vaccine-enhanced

eosino-philia after RSV challenge. To determine whether

immuniza-tion with rAd/3xG potentiates vaccine-enhanced

immuno-pathology, mice were challenged with RSV 3 weeks after

immunization and the levels of eosinophilia in BAL were

ex-amined at 5 days after challenge. As previously reported (36,

40), eosinophilia was markedly enhanced in vvG-immunized

animals (Fig. 4C) (15 to 25% of total BAL fluid cells).

How-ever, a weak infiltrate of eosinophils, ranging from 1 to 3% of

the total BAL fluid cells, was observed in mice previously

immunized with the rAd/3xG vaccine. This influx was similar in

magnitude to that observed in mice previously infected with

live RSV, suggesting that rAd/3xG hardly increases the risk of

development of eosinophil-related lung pathology.

Protective efficacy of rAd/3xG against RSV challenge.

Hav-ing characterized the immune responses, we next wanted to

examine the immune protection against pulmonary RSV

chal-lenge conferred by mucosal and parenteral rAd/3xG

vaccina-FIG. 3. Immunogenicity of rAd/3xG in the presence of existing

vector immunity. The mice were first inoculated i.n. with various doses

of rAd/control as indicated and then after 3 weeks were immunized i.n.

with 5

⫻ 10

7

PFU of rAd/3xG. Anti-RSV IgG antibody titers were

measured by serum ELISA 3 weeks later. Mucosal IgA titers in the

BAL fluid were also measured 3 weeks after immunization. OD,

op-tical density. Error bars indicate the standard deviation.

V

OL

. 82, 2008

PROTECTIVE IMMUNITY AGAINST RSV INDUCED BY rAd VACCINE

2353

on March 16, 2017 by Ewha Woman`s University

http://jvi.asm.org/

(5)

tion. To this end, immune mice were challenged with live RSV

A2 at 4 weeks after immunization. While there was active RSV

replication in the lungs of the control mice, i.n. immunization

with rAd/3xG prevented any detectable RSV replication in the

lungs during the course of infection (Fig. 5A). A group of mice

previously infected with live RSV A2 also exhibited complete

protection against the challenge. However, i.m. immunization

with rAd/3xG resulted in partial protection at the peak of viral

replication (day 4 after challenge) (Fig. 5A). In keeping with

potent lung protection, there was no significant weight loss

upon RSV challenge (Fig. 5B) and no significant disease score

(data not shown) in rAd/3xG-immune mice compared to

vvG-scarified mice. This protective immunity conferred by rAd/3xG

immunization lasted for more than 10 weeks, since RSV

chal-lenge at 10 weeks after immunization resulted in no detectable

virus replication in the lung (Fig. 5C). Together, these results

suggest that a single mucosal rAd/3xG vaccination gives rise to

long-term protective immunity without priming of pathological

CD4 T cells and subsequent vaccine-enhanced disease.

DISCUSSION

In this study, we targeted an RSV G protein fragment as an

immunogen, expressed by replication-defective rAd, to

evalu-ate its vaccine potential. We chose this G protein fragment for

several reasons: (i) the G glycoprotein is one of the major

immunogens among viral proteins in both humans and rodents

(27), (ii) this region contains a conserved B-cell epitope of both

subgroups A and B (2, 37, 41, 43), and (iii) a subunit vaccine

containing this G protein fragment (9, 33) and a plasmid DNA

vaccine expressing secreted G protein (3, 25) were shown to

induced protective immunity in rodents without induction of

lung pathology. The evidence for the exact role of G in

vaccine-enhanced pathology is still contradictory (21, 28, 44). Thus, it

FIG. 4. G-specific CD4 T-cell response and lung eosinophilia in rAd/3xG-immune mice. (A) Mice were immunized with rAd i.n. or with vvG

by scarification and challenged with RSV 3 weeks after immunization. Lung mononuclear cells were prepared from the lungs of the same groups

of mice (n

⫽ 4) 4 days after challenge and stimulated with G(183–195) peptide in the presence of brefeldin A. Cells were stained for CD4, IFN-␥,

and T-cell receptor V

␤14 and analyzed by flow cytometry. Cells gated for CD4 expression are shown in each dot plot, and the percentages represent

the frequency of G-specific IFN-

␥-positive cells. The numbers in parentheses indicate the percentage of total V␤14

cells among CD4

cells. The

results are representative of two independent experiments. (B) Lung mononuclear cells were stimulated in vitro with plate-coated anti-CD3 for

48 h, and the levels of IL-4 in the culture supernatant were determined by sandwich ELISA. One representative from two separate experiments

is shown. (C) BAL was performed 5 days after challenge, cell pellets were differentially stained, and the number of eosinophils was counted. Data

represent means

⫾ standard deviations (n ⫽ 4). The results are representative of three independent experiments.

on March 16, 2017 by Ewha Woman`s University

http://jvi.asm.org/

(6)

is worthwhile to investigate the protective role of RSV

G-specific immunity elicited by a novel Ad-based vaccine. rAd

vectors have been used successfully and safely in many

preclin-ical and clinpreclin-ical settings (reviewed in reference 38).

Replica-tion-defective Ad is an attractive vaccine delivery vehicle since

it has intrinsic properties that activate both the innate and the

adaptive arms of the immune system (11). It has also been

shown to have very high transduction rate (48). Our results

clearly show that immunization with the rAd/3xG vaccine

in-duces strong serum and mucosal antibody responses after a

single dose without priming of potently immunopathogenic

CD4 T cells, which leads to long-term protection from

exper-imental RSV infection without vaccine-enhanced illness. It is

noteworthy that single-dose immunization with rAd/3xG is

suf-ficient for the induction of protective immunity, while three

immunizations with plasmid DNA encoding RSV G were

re-quired in the DNA vaccine regimen (25). This could be relative

advantage of rAd/3xG over other vaccine candidates,

consid-ering the fact that infant vaccinees must be protected from

infection as soon as possible after birth.

The exact mechanisms responsible for the lack of CD4 T-cell

priming and induction of beneficial and protective immunity by

rAd/3xG need further investigation. One possible explanation

could be based on the intrinsic properties of defective rAd

vaccines. A number of reports of studies using rAd-based

vac-cines describe similar findings that Ad vectors induce potent

transgene product-specific antibody and/or CD8 T-cell

re-sponses but no overly potent CD4 T-cell rere-sponses in

experi-mental animal systems (reviewed in reference 38). Second, it

has been proposed that RSV G has an immunomodulatory

effect on both innate and adaptive immunity (7, 32). For

ex-ample, Bukreyev et al. showed that the secreted form of G and

the cysteine-rich region have a positive modulatory effect on

cytotoxic T-lymphocyte function, which is independent of virus

titers and RSV-induced inflammation (7). Thus, it is possible

that secreted G protein fragments expressed by rAd/3xG affect

the priming environment in a way more favorable to beneficial

protective immunity. Together, our results strongly suggest

that RSV G protein could be successfully applied as a

protec-tive immunogen against RSV infection, if appropriate delivery

vectors such as recombinant Ad are employed.

Our study has also shown that the i.n. route of injection is

better than any other route tested with regard to protective

immunity induced by rAd/3xG. The higher levels of

virus-specific mucosal IgA and serum IgG raised by i.n.

immuniza-tion of rAd/3xG seem to be correlated with better protecimmuniza-tion

against RSV challenge (Fig. 2A and B and 5A). These results

are consistent with previous reports that stimulation and

main-tenance of local mucosal antibody may be important in the

generation of effective protection (12, 34). Since the infection

route for RSV is via the respiratory tract, mucosal delivery

might be critical for the development of protective immunity at

the site of infection. In addition, mucosal immunization has

been reported to play an important role in avoiding

interfer-ence from maternal antibodies against the Ad vector (4, 10).

One of the major concerns about Ad-vectored vaccines is that

the induction of immune responses to this common human

pathogen in neonates and young infants is impaired by

mater-nally transferred antibodies (45). We have shown that i.n.

im-munization with rAd/3xG is capable of inducing strong serum

IgG and mucosal IgA responses even in the presence of vector

immunity (Fig. 3). Previously, it has also been shown that

mucosally delivered Ad vectors expressing viral antigen were

capable of inducing neutralizing antibodies and protective

im-munity in neonatal mice even in the presence of maternal

antibodies against the vaccine carrier (46). These results

sug-gest that when delivered through i.n. route, the rAd/3xG

vac-cine might be effective in human populations in which the

majority have preexisting immunity to Ad serotype 5 as a result

of natural exposure. Furthermore, injection of rAd has been

shown to generally induce inflammatory reactions by releasing

proinflammatory cytokines such as IL-6, tumor necrosis factor

alpha, and IL-1 (14), which may compensate for the Th2 bias

FIG. 5. Immune protection from respiratory RSV challenge and

reduced weight loss by vaccination with rAd/3xG. (A) Each group of

immune mice was challenged with 1

⫻ 10

7

PFU RSV at 4 weeks after

immunization, and the level of RSV replication in the lungs was

de-termined by plaque assay during the postchallenge periods. Results are

expressed as the means

⫾ standard errors of the means from four or

five mice for each time point per group. The limit of detection is 200

PFU/gram of lung. (B) The same groups of immune mice were

chal-lenged with RSV and then weighed each day. Results are expressed as

the means

⫾ standard errors of the means from five mice for each

group.

*

, P

⬍ 0.01;

**

, P

⬍ 0.001. (C) Immune mice were challenged

with 1

⫻ 10

6

PFU RSV at 10 weeks after immunization, and the level

of RSV replication was determined at day 4 after infection. N.D., not

detected.

V

OL

. 82, 2008

PROTECTIVE IMMUNITY AGAINST RSV INDUCED BY rAd VACCINE

2355

on March 16, 2017 by Ewha Woman`s University

http://jvi.asm.org/

(7)

of the immature immune system in neonates. In this regard,

delivery of the RSV G protein fragment by an Ad vector as a

neutralizing antigen might have some advantages over the use

of other vehicles, particularly as neonatal vaccines.

In summary, we have provided evidence that a

replication-deficient rAd-based RSV vaccine represents a promising

can-didate for an RSV vaccine. One of the major advantages

as-sociated with this RSV vaccine might be its effectiveness in

inducing long-term protective immunity with a single dose. It is

also possible that the level and/or duration of protection

in-duced by mucosal vaccination with our vaccine candidate could

be further enhanced when given with appropriate adjuvant(s).

We are currently investigating whether the use of various

doses, mixtures with various adjuvants, and/or multiple

admin-istrations of the immunogen by prime-boost regimens could

further improve the efficacy of our vaccine candidate.

ACKNOWLEDGMENTS

We acknowledge the technical support from Sung-A Kim, Min-Ha

Lee, and Ji-Hye Yoon in the completion of this work.

This work was supported by a Korea Research Foundation grant

funded by the Korean government (MOEHRD) (KRF-2006-E00130),

by grant R15-2006-020 from the NCRC program of MOST and

KOSEF through the Center for Cell Signaling and Drug Discovery

Research at Ewha Womans University, and by the Brain Korea 21

project.

REFERENCES

1. Adkins, B., C. Leclerc, and S. Marshall-Clarke. 2004. Neonatal adaptive immunity comes of age. Nat. Rev. Immunol. 4:553–564.

2. Akerlind-Stopner, B., G. Utter, M. A. Mufson, C. Orvell, R. A. Lerner, and

E. Norrby.1990. A subgroup-specific antigenic site in the G protein of respiratory syncytial virus forms a disulfide-bonded loop. J. Virol. 64:5143– 5148.

3. Andersson, C., P. Liljestrom, S. Stahl, and U. F. Power. 2000. Protection against respiratory syncytial virus (RSV) elicited in mice by plasmid DNA immunisation encoding a secreted RSV G protein-derived antigen. FEMS Immunol. Med. Microbiol. 29:247–253.

4. Belyakov, I. M., B. Moss, W. Strober, and J. A. Berzofsky. 1999. Mucosal vaccination overcomes the barrier to recombinant vaccinia immunization caused by preexisting poxvirus immunity. Proc. Natl. Acad. Sci. USA 96: 4512–4517.

5. Bembridge, G. P., R. Garcia-Beato, J. A. Lopez, J. A. Melero, and G. Taylor. 1998. Subcellular site of expression and route of vaccination influence pul-monary eosinophilia following respiratory syncytial virus challenge in BALB/c mice sensitized to the attachment G protein. J. Immunol. 161:2473– 2480.

6. Brandtzaeg, P. 2003. Role of secretory antibodies in the defence against infections. Int. J. Med. Microbiol. 293:3–15.

7. Bukreyev, A., M. E. Serra, F. R. Laham, G. A. Melendi, S. R. Kleeberger,

P. L. Collins, and F. P. Polack.2006. The cysteine-rich region and secreted form of the attachment G glycoprotein of respiratory syncytial virus enhance the cytotoxic T-lymphocyte response despite lacking major histocompatibil-ity complex class I-restricted epitopes. J. Virol. 80:5854–5861.

8. Connors, M., A. B. Kulkarni, P. L. Collins, C. Y. Firestone, K. L. Holmes,

H. C. Morse III, and B. R. Murphy.1992. Resistance to respiratory syncytial virus (RSV) challenge induced by infection with a vaccinia virus recombinant expressing the RSV M2 protein (Vac-M2) is mediated by CD8⫹T cells, while that induced by Vac-F or Vac-G recombinants is mediated by anti-bodies. J. Virol. 66:1277–1281.

9. Corvaia, N., P. Tournier, T. N. Nguyen, J. F. Haeuw, U. F. Power, H. Binz,

and C. Andreoni.1997. Challenge of BALB/c mice with respiratory syncytial virus does not enhance the Th2 pathway induced after immunization with a recombinant G fusion protein, BBG2NA, in aluminum hydroxide. J. Infect. Dis. 176:560–569.

10. Crowe, J. E., Jr. 2001. Influence of maternal antibodies on neonatal immu-nization against respiratory viruses. Clin. Infect. Dis. 33:1720–1727. 11. Ertl, H. C., and Z. Xiang. 1996. Novel vaccine approaches. J. Immunol.

156:3579–3582.

12. Etchart, N., B. Baaten, S. R. Andersen, L. Hyland, S. Y. Wong, and S. Hou. 2006. Intranasal immunisation with inactivated RSV and bacterial adjuvants induces mucosal protection and abrogates eosinophilia upon challenge. Eur. J. Immunol. 36:1136–1144.

13. Falsey, A. R. 1998. Respiratory syncytial virus infection in older persons. Vaccine 16:1775–1778.

14. Ginsberg, H. S., L. L. Moldawer, P. B. Sehgal, M. Redington, P. L. Kilian,

R. M. Chanock, and G. A. Prince.1991. A mouse model for investigating the molecular pathogenesis of adenovirus pneumonia. Proc. Natl. Acad. Sci. USA 88:1651–1655.

15. Glezen, W. P., S. B. Greenberg, R. L. Atmar, P. A. Piedra, and R. B. Couch. 2000. Impact of respiratory virus infections on persons with chronic under-lying conditions. JAMA 283:499–505.

16. Han, L. L., J. P. Alexander, and L. J. Anderson. 1999. Respiratory syncytial virus pneumonia among the elderly: an assessment of disease burden. J. In-fect. Dis. 179:25–30.

17. Hancock, G. E., D. J. Speelman, K. Heers, E. Bortell, J. Smith, and C. Cosco. 1996. Generation of atypical pulmonary inflammatory responses in BALB/c mice after immunization with the native attachment (G) glycoprotein of respiratory syncytial virus. J. Virol. 70:7783–7791.

18. He, T. C., S. Zhou, L. T. da Costa, J. Yu, K. W. Kinzler, and B. Vogelstein. 1998. A simplified system for generating recombinant adenoviruses. Proc. Natl. Acad. Sci. USA 95:2509–2514.

19. Heidema, J., G. J. de Bree, P. M. De Graaff, W. W. van Maren, P.

Hooger-hout, T. A. Out, J. L. Kimpen, and G. M. van Bleek.2004. Human CD8(⫹)

T cell responses against five newly identified respiratory syncytial virus-derived epitopes. J. Gen. Virol. 85:2365–2374.

20. Johnson, T. R., J. E. Johnson, S. R. Roberts, G. W. Wertz, R. A. Parker, and

B. S. Graham.1998. Priming with secreted glycoprotein G of respiratory syncytial virus (RSV) augments interleukin-5 production and tissue eosino-philia after RSV challenge. J. Virol. 72:2871–2880.

21. Johnson, T. R., M. N. Teng, P. L. Collins, and B. S. Graham. 2004. Respi-ratory syncytial virus (RSV) G glycoprotein is not necessary for vaccine-enhanced disease induced by immunization with formalin-inactivated RSV. J. Virol. 78:6024–6032.

22. Johnson, T. R., S. M. Varga, T. J. Braciale, and B. S. Graham. 2004. V␤14

T cells mediate the vaccine-enhanced disease induced by immunization with respiratory syncytial virus (RSV) G glycoprotein but not with formalin-inactivated RSV. J. Virol. 78:8753–8760.

23. Kjerrulf, M., B. Lowenadler, C. Svanholm, and N. Lycke. 1997. Tandem repeats of T helper epitopes enhance immunogenicity of fusion proteins by promoting processing and presentation. Mol. Immunol. 34:599–608. 24. Levine, S., R. Klaiber-Franco, and P. R. Paradiso. 1987. Demonstration that

glycoprotein G is the attachment protein of respiratory syncytial virus. J. Gen. Virol. 68:2521–2524.

25. Li, X., S. Sambhara, C. X. Li, L. Ettorre, I. Switzer, G. Cates, O. James, M.

Parrington, R. Oomen, R. P. Du, and M. Klein.2000. Plasmid DNA encod-ing the respiratory syncytial virus G protein is a promisencod-ing vaccine candidate. Virology 269:54–65.

26. Martinez, I., J. Dopazo, and J. A. Melero. 1997. Antigenic structure of the human respiratory syncytial virus G glycoprotein and relevance of hypermu-tation events for the generation of antigenic variants. J. Gen. Virol. 78:2419– 2429.

27. Murphy, B. R., S. L. Hall, A. B. Kulkarni, J. E. Crowe, Jr., P. L. Collins, M.

Connors, R. A. Karron, and R. M. Chanock.1994. An update on approaches to the development of respiratory syncytial virus (RSV) and parainfluenza virus type 3 (PIV3) vaccines. Virus Res. 32:13–36.

28. Murphy, B. R., A. Sotnikov, P. R. Paradiso, S. W. Hildreth, A. B. Jenson,

R. B. Baggs, L. Lawrence, J. J. Zubak, R. M. Chanock, J. A. Beeler, et al.

1989. Immunization of cotton rats with the fusion (F) and large (G) glyco-proteins of respiratory syncytial virus (RSV) protects against RSV challenge without potentiating RSV disease. Vaccine 7:533–540.

29. Nicholas, J. A., K. L. Rubino, M. E. Levely, E. G. Adams, and P. L. Collins. 1990. Cytolytic T-lymphocyte responses to respiratory syncytial virus: effector cell phenotype and target proteins. J. Virol. 64:4232–4241.

30. Openshaw, P. J., S. L. Clarke, and F. M. Record. 1992. Pulmonary eosino-philic response to respiratory syncytial virus infection in mice sensitized to the major surface glycoprotein G. Int Immunol. 4:493–500.

31. Oscherwitz, J., M. E. Zeigler, T. E. Gribbin, and K. B. Cease. 1999. A V3 loop haptenic peptide sequence, when tandemly repeated, enhances immu-nogenicity by facilitating helper T-cell responses to a covalently linked car-rier protein. Vaccine 17:2392–2399.

32. Polack, F. P., P. M. Irusta, S. J. Hoffman, M. P. Schiatti, G. A. Melendi,

M. F. Delgado, F. R. Laham, B. Thumar, R. M. Hendry, J. A. Melero, R. A. Karron, P. L. Collins, and S. R. Kleeberger.2005. The cysteine-rich region of respiratory syncytial virus attachment protein inhibits innate immunity elicited by the virus and endotoxin. Proc. Natl. Acad. Sci. USA 102:8996– 9001.

33. Power, U. F., H. Plotnicky-Gilquin, T. Huss, A. Robert, M. Trudel, S. Stahl,

M. Uhlen, T. N. Nguyen, and H. Binz.1997. Induction of protective immunity in rodents by vaccination with a prokaryotically expressed recombinant fu-sion protein containing a respiratory syncytial virus G protein fragment. Virology 230:155–166.

34. Singleton, R., N. Etchart, S. Hou, and L. Hyland. 2003. Inability to evoke a long-lasting protective immune response to respiratory syncytial virus

on March 16, 2017 by Ewha Woman`s University

http://jvi.asm.org/

(8)

tion in mice correlates with ineffective nasal antibody responses. J. Virol.

77:11303–11311.

35. Sparer, T. E., S. Matthews, T. Hussell, A. J. Rae, B. Garcia-Barreno, J. A.

Melero, and P. J. Openshaw.1998. Eliminating a region of respiratory syncytial virus attachment protein allows induction of protective immunity without vaccine-enhanced lung eosinophilia. J. Exp. Med. 187:1921–1926. 36. Srikiatkhachorn, A., and T. J. Braciale. 1997. Virus-specific CD8⫹ T

lym-phocytes downregulate T helper cell type 2 cytokine secretion and pulmo-nary eosinophilia during experimental murine respiratory syncytial virus infection. J. Exp. Med. 186:421–432.

37. Sullender, W. M., K. Anderson, and G. W. Wertz. 1990. The respiratory syncytial virus subgroup B attachment glycoprotein: analysis of sequence, expression from a recombinant vector, and evaluation as an immunogen against homologous and heterologous subgroup virus challenge. Virology

178:195–203.

38. Tatsis, N., and H. C. Ertl. 2004. Adenoviruses as vaccine vectors. Mol. Ther.

10:616–629.

39. Taylor, G., E. J. Stott, M. Bew, B. F. Fernie, P. J. Cote, A. P. Collins, M.

Hughes, and J. Jebbett.1984. Monoclonal antibodies protect against respi-ratory syncytial virus infection in mice. Immunology 52:137–142. 40. Tebbey, P. W., M. Hagen, and G. E. Hancock. 1998. Atypical pulmonary

eosinophilia is mediated by a specific amino acid sequence of the attachment (G) protein of respiratory syncytial virus. J. Exp. Med. 188:1967–1972. 41. Trudel, M., F. Nadon, C. Seguin, and H. Binz. 1991. Protection of BALB/c

mice from respiratory syncytial virus infection by immunization with a syn-thetic peptide derived from the G glycoprotein. Virology 185:749–757.

42. Varga, S. M., X. Wang, R. M. Welsh, and T. J. Braciale. 2001. Immuno-pathology in RSV infection is mediated by a discrete oligoclonal subset of antigen-specific CD4(⫹) T cells. Immunity 15:637–646.

43. Varga, S. M., E. L. Wissinger, and T. J. Braciale. 2000. The attachment (G) glycoprotein of respiratory syncytial virus contains a single immunodominant epitope that elicits both Th1 and Th2 CD4⫹ T cell responses. J. Immunol.

165:6487–6495.

44. Vaux-Peretz, F., J. M. Chapsal, and B. Meignier. 1992. Comparison of the ability of formalin-inactivated respiratory syncytial virus, immunopurified F, G and N proteins and cell lysate to enhance pulmonary changes in Balb/c mice. Vaccine 10:113–118.

45. Wang, Y., Z. Xiang, S. Pasquini, and H. C. Ertl. 1997. Immune response to neonatal genetic immunization. Virology 228:278–284.

46. Xiang, Z., Y. Li, G. Gao, J. M. Wilson, and H. C. Ertl. 2003. Mucosally delivered E1-deleted adenoviral vaccine carriers induce transgene prod-uct-specific antibody responses in neonatal mice. J. Immunol. 171:4287– 4293.

47. Yankai, Z., Y. Rong, H. Yi, L. Wentao, C. Rongyue, Y. Ming, L. Taiming, L.

Jingjing, and W. Jie.2006. Ten tandem repeats of beta-hCG 109–118 en-hance immunogenicity and anti-tumor effects of beta-hCG C-terminal pep-tide carried by mycobacterial heat-shock protein HSP65. Biochem. Biophys. Res. Commun. 345:1365–1371.

48. Zhong, L., A. Granelli-Piperno, Y. Choi, and R. M. Steinman. 1999. Recom-binant adenovirus is an efficient and non-perturbing genetic vector for hu-man dendritic cells. Eur. J. Immunol. 29:964–972.

V

OL

. 82, 2008

PROTECTIVE IMMUNITY AGAINST RSV INDUCED BY rAd VACCINE

2357

on March 16, 2017 by Ewha Woman`s University

http://jvi.asm.org/

수치

FIG. 1. Construction and characterization of rAd/3xG vaccine. (A) A shuttle vector was designed as shown in the diagram and constructed by several cloning steps as described in Materials and Methods
FIG. 2. Characterization of humoral immune responses induced by rAd/3xG. (A) BALB/c mice were immunized once with rAd (rAd/1xG, rAd/3xG, or rAd/control) or with live RSV A2 via the indicated routes, and systemic anti-RSV IgG antibody titers were measured b
FIG. 3. Immunogenicity of rAd/3xG in the presence of existing vector immunity. The mice were first inoculated i.n
FIG. 4. G-specific CD4 T-cell response and lung eosinophilia in rAd/3xG-immune mice. (A) Mice were immunized with rAd i.n
+2

참조

관련 문서

attached to a single building in cluster, unregulated magnification without consideration of urban view, insufficient consideration of the quality of

Magnetic field.. • Single particle motion of the plasma.

 Although van der Waals equation is still less accurate at high

There are clearly stronger materials, which is understandable given that CrMnFeCoNi is a single-phase material, but the toughness of this high-entropy alloy exceeds that

• The back pack type vaccine carrier are easier to carry. • The peltier based system helps to maintain the temperature 2-8 degrees required for the vaccine and prevent

Simulation results show that the proposed MAC protocol has a good throughput performance relative to the single channel and dual-channel MAC protocols at

In this study, we investigated the effects of a HF+HS diet in DMM-induced OA mice and found that compared with a HF diet, a HF+HS diet promoted OA. Moreover, our results

Mortality and clinical signs in SD rats about single oral administration after 3 weeks with callus extract ..... Hematological values in SD rats administered orally